Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Methods ; 10(12): 1225-31, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24141495

RESUMEN

Populations of cells create local environments that lead to emergent heterogeneity. This is particularly evident with human pluripotent stem cells (hPSCs): microenvironmental heterogeneity limits hPSC cell fate control. We developed a high-throughput platform to screen hPSCs in configurable microenvironments in which we optimized colony size, cell density and other parameters to achieve rapid and robust cell fate responses to exogenous cues. We used this platform to perform single-cell protein expression profiling, revealing that Oct4 and Sox2 costaining discriminates pluripotent, neuroectoderm, primitive streak and extraembryonic cell fates. We applied this Oct4-Sox2 code to analyze dose responses of 27 developmental factors to obtain lineage-specific concentration optima and to quantify cell line-specific endogenous signaling pathway activation and differentiation bias. We demonstrated that short-term responses predict definitive endoderm induction efficiency and can be used to rescue differentiation of cell lines reticent to cardiac induction. This platform will facilitate high-throughput hPSC-based screening and quantification of lineage-induction bias.


Asunto(s)
Técnicas de Cultivo de Célula , Endodermo/metabolismo , Células Madre Pluripotentes/citología , Animales , Diferenciación Celular , Línea Celular , Linaje de la Célula , Medios de Cultivo Condicionados/química , Células Madre Embrionarias/citología , Perfilación de la Expresión Génica , Humanos , Ratones , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Fenotipo , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal , Factores de Tiempo
2.
J Mol Cell Cardiol ; 80: 56-70, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25528965

RESUMEN

Differentiation of human pluripotent stem cells as embryoid bodies (EBs) has been achieved previously with p38alfa MAPK inhibitors such as SB203580 with moderate efficiency of 10-15%. We synthesized and screened 42 compounds that are 2,4,5-trisubstituted azole analogues of SB203580 for efficient cardiomyocyte differentiation. Our screen identified novel compounds that have similar cardiac differentiation activity as SB203580. However, the cardiac differentiation did not correlate with p38alfa MAPK inhibition, indicating an alternative mechanism in cardiac differentiation. Upon profiling several 2,4,5-trisubstituted azole compounds against a panel of 97 kinases we identified several off targets, among them casein kinases 1 (CK1). The cardiomyogenic activities of SB203580 and its analogues showed a correlation with post mesoderm Wnt/beta-catenin pathway inhibition of CK1 epsilon and delta. These findings united the mechanism of 2,4,5-trisubstituted azole with the current theory of Wnt/beta-catenin regulated pathway of cardiac differentiation. Consequently an efficient cardiomyocyte protocol was developed with Wnt activator CHIR99021 and 2,4,5-trisubstituted azoles to give high yields of 50-70% cardiomyocytes and a 2-fold increase in growth.


Asunto(s)
Quinasa de la Caseína I/antagonistas & inhibidores , Diferenciación Celular/efectos de los fármacos , Imidazoles/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Piridinas/farmacología , Vía de Señalización Wnt/efectos de los fármacos , Animales , Línea Celular , Diseño de Fármacos , Humanos , Imidazoles/síntesis química , Mesodermo/citología , Mesodermo/efectos de los fármacos , Ratones , Organogénesis/efectos de los fármacos , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/síntesis química
3.
Cytotherapy ; 14(3): 274-84, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22136295

RESUMEN

BACKGROUND AIMS: Human embryonic stem cell (hESC)-derived mesenchymal stromal cells (MSC) (hESC-MSC) are an alternative source of MSC to bone marrow (BM)-derived MSC (BM-MSC), which are being investigated in clinical trials for their immunomodulatory potential. hESC-MSC have the advantage of being consistent because each batch can be generated from hESC under defined conditions. In contrast, BM-MSC have a limited proliferative capacity. METHODS: The ability to suppress the proliferation of anti-CD3/CD28-stimulated CD4 (+) T cells by hESC-MSC was compared with adult BM-MSC and neonatal foreskin fibroblast (Fb). RESULTS: hESC-MSC suppress the proliferation of CD4 (+) T cells in both contact and transwell systems, although inhibition is less in the transwell system. hESC-MSC are approximately 2-fold less potent (67 cells/100 T cells) than BM-MSC and Fb (37 and 34 cells/100 T cells, respectively) at suppressing T-cell proliferation by 50% in a transwell [inhibitory concentration(IC)(50)]. The anti-proliferative effect is not contact-dependent but requires the presence of factors such as interferon (IFN)-γ produced by activated T cells. IFN-γ induces the expression of indoleamine-2,3-dioxygenase (IDO) in hESC-MSC, BM-MSC and Fb, contributing to their immunosuppressive property. CONCLUSIONS: The feedback loop between MSC or Fb and activated T cells may limit the immunosuppressive effects of MSC and Fb to sites containing ongoing immunologic or inflammatory responses where activated T cells induce the up-regulation of IDO and immunomodulatory properties of MSC and Fb. These data demonstrate that hESC-MSC may be evaluated further as an allogeneic cell source for therapeutic applications requiring immunosuppression.


Asunto(s)
Células de la Médula Ósea/citología , Retroalimentación Fisiológica , Terapia de Inmunosupresión/métodos , Células Madre Mesenquimatosas/inmunología , Linfocitos T/inmunología , Adipocitos/citología , Adipocitos/inmunología , Células de la Médula Ósea/metabolismo , Antígenos CD28/inmunología , Complejo CD3/inmunología , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Relación Dosis-Respuesta Inmunológica , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Interferón gamma/inmunología , Activación de Linfocitos , Células Madre Mesenquimatosas/metabolismo , Osteoblastos/citología , Osteoblastos/inmunología , Linfocitos T/citología , Linfocitos T/metabolismo
4.
Stem Cell Reports ; 16(1): 182-197, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33306988

RESUMEN

Universal red blood cells (RBCs) differentiated from O-negative human induced pluripotent stem cells (hiPSCs) could find applications in transfusion medicine. Given that each transfusion unit of blood requires 2 trillion RBCs, efficient bioprocesses need to be developed for large-scale in vitro generation of RBCs. We have developed a scalable suspension agitation culture platform for differentiating hiPSC-microcarrier aggregates into functional RBCs and have demonstrated scalability of the process starting with 6 well plates and finally demonstrating in 500 mL spinner flasks. Differentiation of the best-performing hiPSCs generated 0.85 billion erythroblasts in 50 mL cultures with cell densities approaching 1.7 × 107 cells/mL. Functional (oxygen binding, hemoglobin characterization, membrane integrity, and fluctuations) and transcriptomics evaluations showed minimal differences between hiPSC-derived and adult-derived RBCs. The scalable agitation suspension culture differentiation process we describe here could find applications in future large-scale production of RBCs in controlled bioreactors.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Eritrocitos/metabolismo , Células Madre Pluripotentes Inducidas/citología , Diferenciación Celular , Células Cultivadas , Eritrocitos/citología , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Transcriptoma
5.
Stem Cell Res Ther ; 11(1): 347, 2020 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-32771055

RESUMEN

BACKGROUND: Significant developments in stem cell therapy for Parkinson's disease (PD) have already been achieved; however, methods for reliable assessment of dopamine neuron maturation in vivo are lacking. Establishing the efficacy of new cellular therapies using non-invasive methodologies will be critical for future regulatory approval and application. The current study examines the utility of neuroimaging to characterise the in vivo maturation, innervation and functional dopamine release of transplanted human embryonic stem cell-derived midbrain dopaminergic neurons (hESC-mDAs) in a preclinical model of PD. METHODS: Female NIH RNu rats received a unilateral stereotaxic injection of 6-OHDA into the left medial forebrain bundle to create the PD lesion. hESC-mDA cell and sham transplantations were carried out 1 month post-lesion, with treated animals receiving approximately 4 × 105 cells per transplantation. Behavioural analysis, [18F]FBCTT and [18F]fallypride microPET/CT, was conducted at 1, 3 and 6 months post-transplantation and compared with histological characterisation at 6 months. RESULTS: PET imaging revealed transplant survival and maturation into functional dopaminergic neurons. [18F]FBCTT-PET/CT dopamine transporter (DAT) imaging demonstrated pre-synaptic restoration and [18F]fallypride-PET/CT indicated functional dopamine release, whilst amphetamine-induced rotation showed significant behavioural recovery. Moreover, histology revealed that the grafted cells matured differently in vivo producing high- and low-tyrosine hydroxylase (TH) expressing cohorts, and only [18F]FBCTT uptake was well correlated with differentiation. CONCLUSIONS: This study provides further evidence for the value of in vivo functional imaging for the assessment of cell therapies and highlights the utility of DAT imaging for the determination of early post-transplant cell maturation and differentiation of hESC-mDAs.


Asunto(s)
Neuronas Dopaminérgicas , Enfermedad de Parkinson , Tomografía Computarizada por Tomografía de Emisión de Positrones , Animales , Modelos Animales de Enfermedad , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática , Femenino , Neuroimagen , Oxidopamina , Enfermedad de Parkinson/diagnóstico por imagen , Enfermedad de Parkinson/terapia , Ratas
6.
Stem Cells ; 26(6): 1454-63, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18356574

RESUMEN

Future therapeutic applications of differentiated human embryonic stem cells (hESC) carry a risk of teratoma formation by contaminating undifferentiated hESC. We generated 10 monoclonal antibodies (mAbs) against surface antigens of undifferentiated hESC, showing strong reactivity against undifferentiated, but not differentiated hESC. The mAbs did not cross react with mouse fibroblasts and showed weak to no reactivity against human embryonal carcinoma cells. Notably, one antibody (mAb 84) is cytotoxic to undifferentiated hESC and NCCIT cells in a concentration-dependent, complement-independent manner. mAb 84 induced cell death of undifferentiated, but not differentiated hESC within 30 minutes of incubation, and immunoprecipitation of the mAb-antigen complex revealed that the antigen is podocalyxin-like protein-1. Importantly, we observed absence of tumor formation when hESC and NCCIT cells were treated with mAb 84 prior to transplantation into severe combined immunodeficiency mice. Our data indicate that mAb 84 may be useful in eliminating residual hESC from differentiated cells populations for clinical applications. Disclosure of potential conflicts of interest is found at the end of this article.


Asunto(s)
Células Madre Embrionarias/citología , Sialoglicoproteínas/análisis , Animales , Anticuerpos , Anticuerpos Monoclonales , Diferenciación Celular , Línea Celular , Supervivencia Celular , Células Madre Embrionarias/fisiología , Citometría de Flujo , Células HeLa , Humanos , Ratones , Sialoglicoproteínas/inmunología
7.
Stem Cells Transl Med ; 7(10): 709-720, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30063296

RESUMEN

In this study, 50 tri-substituted imidazoles (TIs), which are analogs of the small molecules TA-01 and SB203580, were synthesized and screened for cardiomyogenic activities. Several TIs displayed cardiomyogenic activities when applied during the differentiation from days 3-5. The TIs did not affect the Wnt/ß-catenin pathway during cardiomyogenesis and the likely mechanism of action is through the inhibition of ALK5 of the TGFß pathway. Interestingly, these TIs promoted the neural differentiation of human pluripotent stem cells (hPSCs) with a similar potency to that of the dual SMAD inhibitors SB431542/LDN-193189 when dosed from days 1 to 9. The neural induction activities of the TIs correlated with their ALK5 inhibitory activities. This study reports the discovery of small molecule inhibitors of ALK5, which can promote the differentiation of hPSCs into cardiomyocytes or neural cells depending on the time of dosing, showing potential for the production of clinical-grade cardiac/neural cells for regenerative therapy. Stem Cells Translational Medicine 2018;7:709-720.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Imidazoles/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Receptor Tipo I de Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Benzamidas/farmacología , Dioxoles/farmacología , Humanos , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/metabolismo , Pirazoles/farmacología , Pirimidinas/farmacología , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
8.
J Biomed Mater Res B Appl Biomater ; 106(5): 1887-1896, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-28941021

RESUMEN

Functionalizing medical devices with polypeptides to enhance their performance has become important for improved clinical success. The extracellular matrix (ECM) adhesion protein vitronectin (VN) is an effective coating, although the chemistry used to attach VN often reduces its bioactivity. In vivo, VN binds the ECM in a sequence-dependent manner with heparan sulfate (HS) glycosaminoglycans. We reasoned therefore that sequence-based affinity chromatography could be used to isolate a VN-binding HS fraction (HS9) for use as a coating material to capture VN onto implant surfaces. Binding avidity and specificity of HS9 were confirmed by enzyme-linked immunosorbent assay (ELISA) and surface plasmon resonance (SPR)-based assays. Plasma polymerization of allylamine (AA) to tissue culture-treated polystyrene (TCPS) was then used to capture and present HS9 as determined by radiolabeling and ELISA. HS9-coated TCPS avidly bound VN, and this layered surface supported the robust attachment, expansion, and maintenance of human pluripotent stem cells. Compositional analysis demonstrated that 6-O- and N-sulfation, as well as lengths greater than three disaccharide units (dp6) are critical for VN binding to HS-coated surfaces. Importantly, HS9 coating reduced the threshold concentration of VN required to create an optimally bioactive surface for pluripotent stem cells. We conclude that affinity-purified heparan sugars are able to coat materials to efficiently bind adhesive factors for biomedical applications. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 106B: 1887-1896, 2018.


Asunto(s)
Materiales Biocompatibles Revestidos/química , Proteínas de la Matriz Extracelular/química , Heparitina Sulfato/química , Células Madre Pluripotentes/metabolismo , Vitronectina/química , Adhesión Celular , Línea Celular , Humanos , Células Madre Pluripotentes/citología
9.
Stem Cells Dev ; 16(3): 413-20, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17610371

RESUMEN

We employed a stromal-derived inducing activity (SDIA) model of neurogenesis to investigate the effects of targeted knockdown of Oct-4 and Sox-2 by short interfering RNAs (siRNAs) in mouse embryonic stem (mES) cells. Quantitative real-time PCR showed 40-90% knockdown of specific transcripts with cognate Oct-4 or Sox-2 siRNA transfection compared to FAM-labeled negative control (FAM) siRNA or mock transfection and was confirmed at the protein level by western blot analyses. Upon differentiation using PA6 SDIA co-cultures, neurogenesis is significantly diminished in Oct-4 or Sox-2-targeted mES cells. It was observed that 45 +/- 12%, 65 +/- 13%, and 90 +/- 8% of the colonies were stained with neuron-specific beta-tubulin III in Oct-4, Sox-2, and FAM siRNA transfected mES cells, respectively, with similar results observed using neural inducing factors collected from the surface of PA6. Together, our results extend observations for a role of Oct-4 in SDIA and implicate a similar role for Sox-2.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias/fisiología , Proteínas HMGB/metabolismo , Neuronas/fisiología , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , ARN Interferente Pequeño/metabolismo , Factores de Transcripción/metabolismo , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Técnicas de Cocultivo , Proteínas de Unión al ADN/genética , Células Madre Embrionarias/citología , Regulación de la Expresión Génica , Proteínas HMGB/genética , Ratones , Neuronas/citología , Factor 3 de Transcripción de Unión a Octámeros/genética , ARN Interferente Pequeño/genética , Factores de Transcripción SOXB1 , Factores de Transcripción/genética , Transcripción Genética
10.
Stem Cells Transl Med ; 6(9): 1803-1814, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28650520

RESUMEN

Recent reports have indicated human embryonic stem cells-derived midbrain dopamine (mDA) neurons as proper cell resources for use in Parkinson's disease (PD) therapy. Nevertheless, no detailed and systematic study has been conducted to identify which differentiation stages of mDA cells are most suitable for transplantation in PD therapy. Here, we transplanted three types of mDA cells, DA progenitors (differentiated in vitro for 16 days [D16]), immature DA neurons (D25), and DA neurons (D35), into PD mice and found that all three types of cells showed high viability and strong neuronal differentiation in vivo. Both D25 and D35 cells showed neuronal maturation and differentiation toward TH+ cells and, accordingly, satisfactory behavioral functional recovery. However, transplanted D16 cells were less capable of producing functional recovery. These findings provide a valuable guideline for standardizing the differentiation stage of the transplantable cells used in clinical cell therapy for PD. Stem Cells Translational Medicine 2017;6:1803-1814.


Asunto(s)
Neuronas Dopaminérgicas/citología , Mesencéfalo/citología , Células-Madre Neurales/citología , Enfermedad de Parkinson/terapia , Trasplante de Células Madre/métodos , Animales , Células Cultivadas , Neuronas Dopaminérgicas/trasplante , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células-Madre Neurales/trasplante , Neurogénesis
11.
J Biotechnol ; 122(3): 341-61, 2006 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-16494961

RESUMEN

In this study we report observations that mouse embryonic fibroblasts (MEF) capable of supporting expansion of pluripotent, human embryonic stem cells (hESC) fail to support after immortalization using E6/E7 oncogenes in serum conditions; however this can be reversed following addition of exogenous TGF-beta2. Microarray analysis of immortalized and non-immortalized MEF revealed differential gene expression of several TGF-beta related genes. By supplementing TGF-beta2 into E6/E7 immortalized MEF cultures, this enabled proliferation of undifferentiated, pluripotent hESC as demonstrated by marker expression (Oct-4, SSEA-4, alkaline phosphatase) and teratoma formation representing three germ layers following hESC injection into immuno-deficient mice. Subsequent investigation using quantitative real-time PCR highlighted differential gene expression of several extracellular matrix related transcripts in primary and immortal (+/-TGF-beta2) feeder cells including the induction of osteopontin following addition of TGF-beta2. Our results demonstrate that TGF-beta2 and its related genes in MEF play a role in the support of pluripotent hESC expansion.


Asunto(s)
Células Madre Pluripotentes/citología , Factor de Crecimiento Transformador beta/farmacología , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Embrión de Mamíferos/citología , Fibroblastos/citología , Humanos , Ratones , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/metabolismo , Osteopontina , Proteínas E7 de Papillomavirus/genética , Proteínas E7 de Papillomavirus/metabolismo , Sialoglicoproteínas/biosíntesis , Factor de Crecimiento Transformador beta2
12.
J Biotechnol ; 122(1): 130-41, 2006 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-16233925

RESUMEN

Human embryonic stem cells (hESC) are pluripotent cells that proliferate indefinitely in culture, whilst retaining their capacity for differentiation into different cell types. However, hESC cultures require culture in direct contact with feeder cells or conditioned medium (CM) from feeder cells. The most common source of feeders has been primary mouse embryonic fibroblast (MEF). In this study, we immortalized a primary MEF line with the E6 and E7 genes from HPV16. The immortal line, DeltaE-MEF, was able to proliferate beyond 7-9 passages and has an extended lifespan beyond 70 passages. When tested for its ability to support hESC growth, it was found that hESC continue to maintain the undifferentiated morphology for >40 passages both in co-culture with DeltaE-MEF and in feeder-free cultures supplemented with CM from DeltaE-MEF. The cultures also continue to express the pluripotent markers, Oct-4, SSEA-4, Tra-1-60, Tra-1-81, alkaline phosphatase and maintain a normal karyotype. In addition, these hESC formed teratomas when injected into SCID mice. Lastly, we demonstrated the feasibility of scaling-up significant quantities of undifferentiated hESC (>10(8) cells) using DeltaE-MEF in cell factories. The results from this study suggest that immortalized feeders can provide a consistent and reproducible source of feeders for hESC expansion and research.


Asunto(s)
Sistema Libre de Células/metabolismo , Técnicas de Cocultivo/métodos , Fibroblastos/citología , Fibroblastos/fisiología , Células Madre/citología , Células Madre/fisiología , Animales , Comunicación Celular/fisiología , Diferenciación Celular , Línea Celular , Proliferación Celular , Tamaño de la Célula , Supervivencia Celular , Humanos , Ratones , Ratones SCID , Proyectos Piloto
13.
Cell Transplant ; 25(7): 1343-57, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26720780

RESUMEN

Neuronal progenitor cells (NPCs) derived from human embryonic stem cells (hESCs) are an excellent cell source for transplantation therapy due to their availability and ethical acceptability. However, the traditional method of expansion and differentiation of hESCs into NPCs in monolayer cultures requires a long time, and the cell yield is low. A microcarrier (MC) platform can improve the expansion of hESCs and increase the yield of NPCs. In this study, for the first time, we transplanted microcarrier-expanded hESC-derived NPCs into the striatum of adult NOD-SCID IL2Rgc null mice, either as single cells or as cell aggregates. The recipient mice were perfused, and the in vivo survival, differentiation, and targeted innervation of the transplanted cells were assessed by immunostaining. We found that both the transplanted single NPCs and aggregate NPCs were able to survive 1 month posttransplantation, as revealed by human-specific neural cell adhesion molecule (NCAM) and human nuclear antigen staining. Compared to the single cells, the transplanted cell aggregates showed better survival over a 3-month period. In addition, both the transplanted single NPCs and the aggregate NPCs were able to differentiate into DCX-positive immature neurons and Tuj1-positive neurons in vivo by 1 month posttransplantation. However, only the transplantation of aggregate NPCs was shown to result in mature neurons at 3 months posttransplantation. Furthermore, we found that the cell aggregates were able to send long axons to innervate their targets. Our study provides preclinical evidence that the use of MCs to expand and differentiate hESC-derived NPCs and transplantation of these cells as aggregates produce longer survival in vivo.


Asunto(s)
Diferenciación Celular , Microesferas , Células-Madre Neurales/citología , Células-Madre Neurales/trasplante , Animales , Agregación Celular , Proliferación Celular , Supervivencia Celular , Neuronas Dopaminérgicas/citología , Proteína Doblecortina , Células Madre Embrionarias Humanas/citología , Humanos , Masculino , Ratones Endogámicos NOD , Ratones SCID , Neuritas/metabolismo , Factores de Tiempo
14.
Tissue Eng Part A ; 21(9-10): 1507-19, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25602926

RESUMEN

Functional vascularization is a prerequisite for cardiac tissue engineering of constructs with physiological thicknesses. We previously reported the successful preservation of main vascular conduits in isolated thick acellular porcine cardiac ventricular ECM (pcECM). We now unveil this scaffold's potential in supporting human cardiomyocytes and promoting new blood vessel development ex vivo, providing long-term cell support in the construct bulk. A custom-designed perfusion bioreactor was developed to remodel such vascularization ex vivo, demonstrating, for the first time, functional angiogenesis in vitro with various stages of vessel maturation supporting up to 1.7 mm thick constructs. A robust methodology was developed to assess the pcECM maximal cell capacity, which resembled the human heart cell density. Taken together these results demonstrate feasibility of producing physiological-like constructs such as the thick pcECM suggested here as a prospective treatment for end-stage heart failure. Methodologies reported herein may also benefit other tissues, offering a valuable in vitro setting for "thick-tissue" engineering strategies toward large animal in vivo studies.


Asunto(s)
Matriz Extracelular/metabolismo , Miocardio/metabolismo , Neovascularización Fisiológica , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Animales , Reactores Biológicos , Técnicas de Cocultivo , Estudios de Factibilidad , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Células Madre Mesenquimatosas/citología , Miocardio/citología , Sus scrofa
15.
Regen Med ; 8(4): 413-24, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23826696

RESUMEN

BACKGROUND: Human embryonic stem cells (hESCs) have been derived and maintained on mouse embryonic fibroblast feeders to keep their undifferentiated status. To realize their clinical potential, a feeder-free and scalable system for large scale production of hESCs and their differentiated derivatives is required. MATERIALS & METHODS: hESCs were cultured and passaged on serum/feeder-free 3D microcarriers for five passages. For embryoid body (EB) formation and hemangioblast differentiation, the medium for 3D microcarriers was directly switched to EB medium. RESULTS: hESCs on 3D microcarriers maintained pluripotency and formed EBs, which were ten-times more efficient than hESCs cultured under 2D feeder-free conditions (0.11 ± 0.03 EB cells/hESC input 2D vs 1.19 ± 0.32 EB cells/hESC input 3D). After replating, EB cells from 3D culture readily developed into hemangioblasts with the potential to differentiate into hematopoietic and endothelial cells. Furthermore, this 3D system can also be adapted to human induced pluripotent stem cells, which generate functional hemangioblasts with high efficiency. CONCLUSION: This 3D serum- and stromal-free microcarrier system is important for future clinical applications, with the potential of developing to a GMP-compatible scalable system.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Medio de Cultivo Libre de Suero , DEAE-Celulosa/química , Células Nutrientes , Hemangioblastos/citología , Células Madre Hematopoyéticas/citología , Células Madre Pluripotentes/citología , Animales , Células Cultivadas , Colágeno/metabolismo , Combinación de Medicamentos , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Hemangioblastos/metabolismo , Células Madre Hematopoyéticas/metabolismo , Humanos , Laminina/metabolismo , Ratones , Microesferas , Células Madre Pluripotentes/metabolismo , Proteoglicanos/metabolismo
16.
Tissue Eng Part C Methods ; 19(2): 166-80, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22834957

RESUMEN

Neural progenitor cells (NPCs) derived from human induced pluripotent stem cells (hiPSCs) can be differentiated to neural cells that model neurodegenerative diseases and be used in the screening of potential drugs to ameliorate the disease phenotype. Traditionally, NPCs are produced in 2D cultures, in low yields, using a laborious process that includes generation of embryonic bodies, plating, and colony selections. To simplify the process and generate large numbers of hiPSC-derived NPCs, we introduce a microcarrier (MC) system for the expansion of a hiPSC line and its subsequent differentiation to NPC, using iPS (IMR90) as a model cell line. In the expansion stage, a process of cell propagation in serum-free MC culture was developed first in static culture, which is then scaled up in stirred spinner flasks. A 7.7-fold expansion of iPS (IMR90) and cell yield of 1.3×106 cells/mL in 7 days of static MC culture were achieved. These cells maintained expression of OCT 3/4 and TRA-1-60 and possessed a normal karyotype over 10 passages. A higher cell yield of 6.1×106 cells/mL and 20-fold hiPSC expansion were attained using stirred spinner flasks (seeded from MC static cultures) and changing the medium-exchange regimen from once to twice a day. In the differentiation stage, NPCs were generated with 78%-85% efficiency from hiPSCs using a simple serum-free differentiation protocol. Finally, the integrated process of cell expansion and differentiation of hiPSCs into NPCs using an MC in spinner flasks yielded 333 NPCs per seeded hiPSC as compared to 53 in the classical 2D tissue culture protocol. Similar results were obtained with the HES-3 human embryonic stem cell line. These NPCs were further differentiated into ßIII-tubulin⁺ neurons, GFAP⁺ astrocytes, and O4⁺ oligodendrocytes, showing that cells maintained their multilineage differentiation potential.


Asunto(s)
Diferenciación Celular , División Celular , Neuronas/citología , Células Madre Pluripotentes/citología , Células Cultivadas , Medio de Cultivo Libre de Suero , Citometría de Flujo , Humanos , Inmunohistoquímica , Cariotipificación , Reacción en Cadena en Tiempo Real de la Polimerasa
17.
J Cardiovasc Transl Res ; 6(6): 989-99, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24081385

RESUMEN

While human embryonic stem cells (hESCs) can differentiate into functional cardiomyocytes, their immature phenotypes limit their therapeutic application for myocardial regeneration. We sought to determine whether electrical stimulation could enhance the differentiation and maturation of hESC-derived cardiomyocytes. Cardiac differentiation was induced in a HES3 hESC line via embryoid bodies formation treated with a p38 MAP kinase inhibitor. Detailed molecular and functional analysis were performed in those hESC-derived cardiomyocytes cultured for 4 days in the absence or presence of electrical field stimulation (6.6 V/cm, 1 Hz, and 2 ms pulses) using an eight-channel C-Pace stimulator (Ion-Optics Co., MA). Upon electrical stimulation, quantitative polymerase chain reaction demonstrated significant upregulation of cardiac-specific gene expression including HCN1, MLC2V, SCN5A, SERCA, Kv4.3, and GATA4; immunostaining and flow cytometry analysis revealed cellular elongation and an increased proportion of troponin-T positive cells (6.3 ± 1.2% vs. 15.8 ± 2.1%; n = 3, P < 0.01). Electrophysiological studies showed an increase in the proportion of ventricular-like hESC-derived cardiomyocytes (48 vs. 29%, P < 0.05) with lengthening of their action potential duration at 90% repolarization (387.7 ± 35.35; n = 11 vs. 291.8 ± 20.82; n = 10, P < 0.05) and 50% repolarization (313.9 ± 27.94; n = 11 vs. 234.0 ± 16.10; n = 10, P < 0.05) after electrical stimulation. Nonetheless, the membrane diastolic potentials and action potential upstrokes of different hESC-derived cardiomyocyte phenotypes, and the overall beating rate remained unchanged (all P > 0.05). Fluorescence confocal imaging revealed that electrical stimulation significantly increased both spontaneous and caffeine-induced calcium flux in the hESC-derived cardiomyocytes (approximately 1.6-fold for both cases; P < 0.01). In conclusion, electrical field stimulation increased the expression of cardiac-specific genes and the yield of differentiation, promoted ventricular-like phenotypes, and improved the calcium handling of hESC-derived cardiomyocytes.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/fisiología , Miocitos Cardíacos/fisiología , Señalización del Calcio , Línea Celular , Linaje de la Célula , Estimulación Eléctrica , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/enzimología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Potenciales de la Membrana , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/enzimología , Fenotipo , Inhibidores de Proteínas Quinasas/farmacología , Factores de Tiempo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
18.
Tissue Eng Part C Methods ; 17(2): 165-72, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20698747

RESUMEN

One of the factors that can impact human embryonic stem cell expansion in stirred microcarrier culture reactors is mechanical stress caused by agitation. Therefore, we have investigated the effects of agitation on human embryonic stem cell growth and expression of pluripotent markers. Agitation of HES-2 cell line in microcarrier cultures in stirred spinner and agitated six-well plates did not affect expression of pluripotent markers, cell viability, and cell doubling times even after seven passages. However, HES-3 cell line was found to be shear sensitive, showing downregulation of three pluripotent markers Oct-4, mAb 84, and Tra-1-60, and lower cell densities in agitated as compared with static cultures, even after one passage. Cell viability was unaffected. The HES-3-agitated cultures showed increased expression of genes and proteins of the three germ layers. We were unable to prevent loss of pluripotent markers or restore doubling times in agitated HES-3 microcarrier cultures by addition of five different known cell protective polymers. In addition, the human induced pluripotent cell line IMR90 was also shown to differentiate in agitated conditions. These results indicate that the effect of agitation on cell growth and differentiation is cell line specific. We assume that the changes in the growth and differentiation of the agitation-sensitive (HES-3) cell line do not result from the effect of shear stress directly on cell viability, but rather by signaling effects that influence the cells to differentiate resulting in slower growth.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Células Madre Pluripotentes/citología , Estrés Mecánico , Diferenciación Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Peso Molecular , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/metabolismo , Polímeros/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Tiempo
19.
Tissue Eng Part C Methods ; 17(2): 193-207, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20726687

RESUMEN

Current methodology for pluripotent human embryonic stem cells (hESCs) expansion relies on murine sarcoma basement membrane substrates (Matrigel™), which precludes the use of these cells in regenerative medicine. To realize the clinical efficacy of hESCs and their derivatives, expansion of these cells in a defined system that is free of animal components is required. This study reports the successful propagation of hESCs (HES-3 and H1) for > 20 passages on tissue culture-treated polystyrene plates, coated from 5 µg/mL of human plasma-purified vitronectin (VN) solution. Cells maintain expression of pluripotent markers Tra1-60 and OCT-4 and are karyotypically normal after 20 passages of continuous culture. In vitro and in vivo differentiation of hESC by embryoid body formation and teratoma yielded cells from the ecto-, endo-, and mesoderm lineages. VN immobilized on tissue culture polystyrene was characterized using a combination of X-ray photoemission spectroscopy, atomic force microscopy, and quantification of the VN surface density with a Bradford protein assay. Ponceau S staining was used to measure VN adsorption and desorption kinetics. Tuning the VN surface density, via the concentration of depositing solution, revealed a threshold surface density of 250 ng/cm², which is required for hESCs attachment, proliferation, and differentiation. Cell attachment and proliferation assays on VN surface densities above this threshold show the substrate properties to be equally viable.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Vitronectina/farmacología , Adsorción/efectos de los fármacos , Biomarcadores/metabolismo , Adhesión Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Células Madre Embrionarias/metabolismo , Humanos , Análisis Espectral , Propiedades de Superficie/efectos de los fármacos , Factores de Tiempo
20.
Methods Mol Biol ; 650: 75-83, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20686944

RESUMEN

The following protocols provide a rapid approach for establishing good working conditions for transfecting siRNAs for specific gene knockdown. By first using microscopy to evaluate efficient transfection of an inexpensive, fluorescent oligonucleotide, the researcher can later proceed with more expensive Western blot or quantitative real-time PCR (qRT-PCR) methods. Thus, the main culprit of ineffective knockdown, poor transfection, can be eliminated before engaging in tedious and time-consuming approaches for troubleshooting siRNA knockdown experiments.


Asunto(s)
Células Madre Embrionarias/metabolismo , Técnicas de Silenciamiento del Gen/métodos , Oligonucleótidos/genética , ARN Interferente Pequeño/genética , Animales , Línea Celular , Ratones , Microscopía Fluorescente , Interferencia de ARN/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA